WO2018217558A1 - Composés de 5-méthyl-1,3,4-oxadiazol-2-yl - Google Patents

Composés de 5-méthyl-1,3,4-oxadiazol-2-yl Download PDF

Info

Publication number
WO2018217558A1
WO2018217558A1 PCT/US2018/033368 US2018033368W WO2018217558A1 WO 2018217558 A1 WO2018217558 A1 WO 2018217558A1 US 2018033368 W US2018033368 W US 2018033368W WO 2018217558 A1 WO2018217558 A1 WO 2018217558A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
methyl
pharmaceutically acceptable
acceptable salt
mixture
Prior art date
Application number
PCT/US2018/033368
Other languages
English (en)
Inventor
Nicolas Jacques Francois Dreyfus
Peter James LINDSAY-SCOTT
Richard Edmund Rathmell
Original Assignee
Eli Lilly And Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Priority to CN201880033585.3A priority Critical patent/CN110650957B/zh
Priority to EP18729282.6A priority patent/EP3630755B1/fr
Priority to ES18729282T priority patent/ES2935349T3/es
Priority to JP2019564808A priority patent/JP6810285B2/ja
Publication of WO2018217558A1 publication Critical patent/WO2018217558A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/09Geometrical isomers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the present invention relates to novel 5-methyl-l,3,4-oxadiazol-2-yl compounds, to pharmaceutical compositions comprising the compounds, to methods of using the compounds to treat physiological disorders, and to intermediates and processes useful in the synthesis of the compounds.
  • the present invention is in the field of treatment of Alzheimer's disease, progressive supranuclear palsy (PSP) and other diseases and disorders involving tau-mediated neurodegeneration, known collectively as tauopathies.
  • PSP progressive supranuclear palsy
  • tauopathies other diseases and disorders involving tau-mediated neurodegeneration, known collectively as tauopathies.
  • Alzheimer's disease is a devastating neurodegenerative disorder that affects millions of patients worldwide.
  • agents on the market which afford only transient, symptomatic benefits to the patient, there is a significant unmet need in the treatment of Alzheimer's disease.
  • the oligomerization of the microtubule-associated protein tau into filamentous structures such as paired helical filaments (PHFs) and straight or twisted filaments, which give rise to neurofibrillary tangles (NFTs) and neuropil threads (NTs), is one of the defining pathological features of Alzheimer's disease and other tauopathies.
  • the number of NFTs in the brains of individuals with Alzheimer's disease has been found to correlate closely with the severity of the disease, suggesting tau has a key role in neuronal dysfunction and neurodegeneration (Nelson et al., J Neuropathol Exp Neurol, 71(5), 362- 381(2012)).
  • Tau pathology has been shown to correlate with disease duration in PSP; cases with a more aggressive disease course have a higher tau burden than cases with a slower progression. (Williams et al., Brain, 130, 1566-76 (2007)).
  • OGA O-GlcNAcase
  • OGA inhibitors are recognized as a valid therapeutic approach to reduce the accumulation of hyperphosphorylated, pathological forms of tau.
  • U. S. Patent No. 9, 120,781 discloses hexahydrobenzooxazole and hexahydrobenzothiazole derivatives which possess OGA inhibitory activity and are further disclosed as useful in treating diseases and disorders related to deficiency or overexpression of OGA, and/or accumulation or deficiency of 2-acetamido-2-deoxy-5B- D-glucopyranoside (O-GlcNAc).
  • O-GlcNAc 2-acetamido-2-deoxy-5B- D-glucopyranoside
  • US 2016/0031871 discloses certain glycosidase inhibitors for treating Alzheimer's disease.
  • OGA inhibitors that are brain penetrant are desired to provide treatments for tau-mediated neurodegeneration disorders, such as Alzheimer's disease and PSP.
  • the present invention provides certain novel compounds that are inhibitors of OGA.
  • the present invention also provides a method of treating Alzheimer's disease in a patient in need of such treatment, comprising administering to the patient an effective amount of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof.
  • the present invention further provides a method of treating the progression of mild cognitive impairment to Alzheimer' s disease in a patient in need of such treatment, comprising administering to the patient an effective amount of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof.
  • the present invention also provides a method of treating progressive supranuclear palsy in a patient in need of such treatment, comprising administering to the patient an effective amount of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof.
  • the present invention also provides a method of treating tau-mediated neurodegenerative disorders in a patient, comprising administering to a patient in need of such treatment an effective amount of a compound of Formulas I or la, or a
  • this invention provides a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof for use in therapy, in particular for use in the treatment of Alzheimer's disease or for use in preventing the progression of mild cognitive impairment to Alzheimer's disease.
  • this invention provides a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof for use in the treatment of progressive supranuclear palsy.
  • the invention also provides a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof for use in treating tau- mediated neurodegenerative disorders.
  • this invention provides the use of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of Alzheimer's disease or for preventing the progression of mild cognitive impairment to Alzheimer's disease.
  • this invention provides the use of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of progressive supranuclear palsy.
  • the invention also provides the use of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for treating tau-mediated neurodegenerative disorders.
  • the invention further provides a pharmaceutical composition, comprising a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • the invention further provides a process for preparing a pharmaceutical composition, comprising admixing a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • This invention also encompasses novel intermediates and processes for the synthesis of the compounds of Formulas I and la. For example, the invention further provides the following
  • Suitable protecting groups include tert-butyl carboxylate and the like.
  • the invention also provides an intermediate compound of Formula Ila:
  • Mild cognitive impairment has been defined as a potential prodromal phase of dementia associated with Alzheimer's disease based on clinical presentation and on progression of patients exhibiting mild cognitive impairment to Alzheimer's dementia over time.
  • the term "preventing the progression of mild cognitive impairment to Alzheimer's disease” includes restraining, slowing, stopping, or reversing the progression of mild cognitive impairment to Alzheimer's disease in a patient.
  • treating includes restraining, slowing, stopping, or reversing the progression or severity of an existing symptom or disorder.
  • the term "patient” refers to a human.
  • the term "effective amount” refers to the amount or dose of compound of the invention, or a pharmaceutically acceptable salt thereof which, upon single or multiple dose administration to the patient, provides the desired effect in the patient under diagnosis or treatment.
  • an effective amount can be readily determined by one skilled in the art by the use of known techniques and by observing results obtained under analogous circumstances.
  • determining the effective amount for a patient a number of factors are considered, including, but not limited to: the species of patient; its size, age, and general health; the specific disease or disorder involved; the degree of or involvement or the severity of the disease or disorder; the response of the individual patient; the particular compound administered; the mode of administration; the bioavailability characteristics of the preparation administered; the dose regimen selected; the use of concomitant medication; and other relevant circumstances.
  • the compounds of the present invention are effective at a dosage per day that falls within the range of about 0.1 to about 15 mg/kg of body weight. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed with acceptable side effects, and therefore the above dosage range is not intended to limit the scope of the invention in any way.
  • the compounds of the present invention are formulated as pharmaceutical compositions administered by any route which makes the compound bioavailable, including oral and transdermal routes. Most preferably, such compositions are for oral administration.
  • Such pharmaceutical compositions and processes for preparing same are well known in the art (See, e.g., Remington: The Science and Practice of Pharmacy, L.V. Allen, Editor, 22 nd Edition, Pharmaceutical Press, 2012).
  • Compounds of the present invention include:
  • N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,3,4- oxadiazol-2-yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide is especially preferred.
  • the crystalline form of N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,3,4- oxadiazol-2-yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide which is
  • a peak in the X-ray powder diffraction spectrum at diffraction angle 2- theta of 13.5° in combination with one or more peaks selected from the group consisting of 5.8°, 13.0°, 14.3°, 17.5°, 20.4°, 21.4°, and 22.2° with a tolerance for the diffraction angles of 0.2 degrees, is further preferred.
  • a pharmaceutically acceptable salt of the compounds of the invention can be formed, for example, by reaction of an appropriate free base of a compound of the invention and an appropriate pharmaceutically acceptable acid in a suitable solvent under standard conditions well known in the art.
  • the formation of such salts is well known and appreciated in the art. See, for example, Gould, P.L., "Salt selection for basic drugs," International Journal of Pharmaceutics, 33: 201-217 (1986); Bastin, R.J., et al. "Salt Selection and Optimization Procedures for Pharmaceutical New Chemical Entities,"
  • the compounds of the present invention, or salts thereof may be prepared by a variety of procedures known to one of ordinary skill in the art, some of which are illustrated in the schemes, preparations, and examples below.
  • One of ordinary skill in the art recognizes that the specific synthetic steps for each of the routes described may be combined in different ways, or in conjunction with steps from different schemes, to prepare compounds of the invention, or salts thereof.
  • the products of each step in the schemes below can be recovered by conventional methods well known in the art, including extraction, evaporation, precipitation, chromatography, filtration, trituration, and crystallization. In the schemes below, all substituents unless otherwise indicated, are as previously defined.
  • the reagents and starting materials are readily available to one of ordinary skill in the art.
  • a compound of formula la may be prepared from a compound of formula Ila (Scheme 1). More specifically, a compound of formula Ila is reductively alkylated with N-(4-fluoro-5-formylthiazol-2-yl)acetamide in the presence of a suitable reducing agent such as sodium triacetoxyborohydride in a suitable solvent to provide a compound of formula la. Suitable solvents include ethyl acetate. N-(4-fluoro-5- formylthiazol-2-yl)acetamide may be prepared by methods known in the chemical arts as well as methods provided in the Preparations and Examples.
  • a compound of formula Ila may be prepared from a compound of formula Ilia where Pg is a suitable amine protecting group. More specifically, a compound of formula Ilia where Pg is tert-butyl carboxylate (t-BOC) is reacted with an acid such as
  • a compound of formula Ila may be isolated as the free base or as an acid addition salt corresponding to the acid reagent utilized.
  • Suitable amine protecting groups are known in the chemical arts and include t-BOC and Cbz as well as those discussed in T. W. Green, P. G. M. Wuts, "Protective Groups in Organic Synthesis” Wiley-Interscience, New York, 1999.
  • a compound of formula Ilia where Pg is a suitable amine protecting group may be prepared from a compound of formula IVa (Scheme 2). More specifically, a compound of formula IVa where Pg is tert-butyl carboxylate is reacted with 2-(chloromethyl)-5- methyl-l,3,4-oxadiazole in the presence of a base such as sodium tert-butoxide to provide a compound of formula Ilia. The reaction is conveniently carried out in a solvent such as acetonitrile or dimethylformamide.
  • a compound of formula IVa where Pg is tert-butyl carboxylate may be prepared by reducing a compound of formula Va under hydride or enzymatic conditions. More specifically, a compound of formula Va is reacted with a hydride reducing agent such as lithium tri(sec-butyl)borohydride in a solvent such as tetrahydrofuran to provide a compound of formula IVa where Pg is tert-butyl carboxylate. Alternatively, a compound of formula Va is reacted with an enzymatic reducing agent such as ketoreductase in a solvent such as DMSO to provide a compound of formula IV where Pg is tert-butyl carboxylate.
  • a compound of formula Va where Pg is a suitable amine protecting group may be prepared by processes known in the chemical arts including those described in WO 2004/094380 Al .
  • a compound of formula Ilia where Pg is a suitable amine protecting group may alternatively be prepared from a compound of formula Via in a sequence of steps shown in Scheme 3. More specifically, a compound of formula Via where Pg is tert-butyl carboxylate is reacted with p-toluenesulfonyl chloride in the presence of a base such as diisopropylethylamine to provide a compound of formula Ilia. The reaction is conveniently carried out in a solvent such as acetonitrile.
  • a compound of formula Via where Pg is tert-butyl carboxylate may be prepared by reacting a compound of formula Vila with acetohydrazide in the presence of 1, -carbonyldiimidazole. The reaction is conveniently carried out in a solvent such as tetrahydrofuran.
  • a compound of formula Vila where Pg is tert-butyl carboxylate may be prepared by first reacting a compound of formula IVa with 2-chloro-l-morpholino-ethanone and a base such as sodium tert- butoxide in a solvent such as acetonitrile.
  • the resulting morpholino adduct of a compound of formula IVa is then hydrolysed with an aqueous base such as aqueous sodium hydroxide in a solvent such as 2-propanol to provide a compound of formula Vila where Pg is tert-butyl carboxylate.
  • an aqueous base such as aqueous sodium hydroxide in a solvent such as 2-propanol to provide a compound of formula Vila where Pg is tert-butyl carboxylate.
  • Cesium fluoride (227 g, 1480 mmol) is added to a solution of tert-butyl N-(4- chloro-5-formyl-thiazol-2-yl)carbamate (38.8 g, 148 mmol; for preparation of tert-butyl N-(4-chloro-5-formyl-thiazol-2-yl)carbamate see for example, N. Masuda, et al., Bioorg Med Chem, 12, 6171-6182 (2004)) in DMSO (776 mL) at room temperature. The reaction mixture is stirred in a 145 °C heating block with an internal temperature of 133 °C for 48 hours, then the mixture is cooled in an ice-water bath.
  • the jacket temperature is then set to -30 °C and pyridine (110 mL, 1360 mmol) is added dropwise over 5 minutes, maintaining an internal temperature below 5 °C.
  • the jacket temperature is set to 0 °C and acetic anhydride (116 mL, 1220 mmol) is added dropwise over 5 minutes.
  • the reaction mixture is stirred overnight at an internal temperature of 37 °C, then is cooled to room temperature and passed through a short pad of diatomaceous earth, eluting with tetrahydrofuran (500 mL).
  • the filtrate is transferred to a flask and the mixture is concentrated to give a residue, which is concentrated from toluene (50 mL).
  • tert-butyl (2S)-2-methyl-4-oxo-piperidine-l-carboxylate 50 g, 234.44 mmol
  • tetrahydrofuran 500 mL
  • the mixture is cooled to -65 °C under an atmosphere of nitrogen and lithium tri(sec-butyl)borohydride (304.77 mL, 304.77 mmol; 1 M in tetrahydrofuran) is added dropwise over 45 minutes, maintaining an internal temperature below -60 °C.
  • the reaction mixture is stirred at room temperature for 1 hour, then is cooled to -30 °C.
  • dihydrogen phosphate (6.5 kg, 0.41 equiv) at 20 °C is charged DMSO (27.4 kg , 1.0 vol) and D-(+)-glucose monohydrate (28.9 kg, 1.25 equiv).
  • the internal temperature is adjusted to 30 °C, and the pH of the reaction is adjusted to 6.9 by addition of aqueous sodium hydroxide (8%, 15 L, 0.28 equiv).
  • the reactor is charged with tert-butyl (2S)-2- methyl-4-oxo-piperidine-l-carboxylate (24.9 kg, 1.0 equiv (99.1%ee)), and the mixture is agitated at 30 °C for 15 min.
  • Ketoreductase (KRED-130, 250 g, 1% w/w), glucose dehydrogenase (GDH-101, 250 g, 1% w/w), and NADP sodium salt (63 g, 0.25% w/w) are charged directly to the reaction mixture via an open port.
  • the mixture is maintained at a temperature of 30 °C and pH 7.0 ⁇ 0.2 via addition of 8% aqueous NaHC0 3 .
  • the reaction is charged with CeliteTM (12.5 kg, 50 w/w%) and toluene (125 L, 5 vol).
  • the mixture is transferred to another 2000 L reactor via an in-line GAF-filter (4 sock) over the period of 1 h.
  • the mixture is allowed to stand 30 min without agitation, the layers are separated, and the aqueous layer is back-extracted with toluene (2 x 125 L).
  • the combined organic layers are filtered (in-line GAF-filter), and the toluene mixture is washed with aqueous sodium chloride solution (25%, 125 L, 5 vol) at 25 °C.
  • the resulting toluene solution is azeotropically dried (partial vacuum, internal temp ⁇ 60 °C) to 0.10 w/w% water, and cooled to 20 °C.
  • the mixture is filtered out of the reactor via a cartridge filter into clean drums under positive nitrogen pressure.
  • the reaction mixture is then transferred from the drums into a 500 L glass lined vessel and concentrated under vacuum ( ⁇ 60 °C) to a target residual volume of 56 L (2.25 vol).
  • n-Heptane (169 kg, 10 vol) is charged at 40 °C, and the mixture is seeded with 25 g of tert-butyl (2S,4S)-4-hydroxy-2-m ethyl -piperi dine- 1- carboxylate.
  • the resulting thick slurry is diluted with additional n-heptane (25 L, 1 vol) and cooled to 16 °C over 4 h.
  • 2-Chloro-l-morpholino-ethanone (59.4 g, 363 mmol) is added to a solution of tert-butyl (2S,4S)-4-hydroxy-2-methyl-piperidine-l-carboxylate (52.1 g, 242 mmol) in acetonitrile (521 mL) at room temperature.
  • the reaction mixture is stirred in an ice-water bath and sodium tert-butoxide (48.0 g, 484 mmol) is added in portions over 10 minutes, maintaining an internal temperature below 15 °C.
  • the reaction mixture is stirred at room temperature for 2 hours, then is added over 5 minutes to another flask containing saturated aqueous ammonium chloride solution (250 mL) and water (250 mL) with ice-water bath cooling, maintaining an internal temperature below 15 °C during the addition.
  • the mixture is warmed to room temperature and extracted with methyl tert- butyl ether (2 x 500 mL), then the combined organics are washed with brine (300 mL).
  • the combined organics are then dried over sodium sulfate, filtered, and concentrated to give a residue, which is combined with 2-propanol (414 mL) and 2M aqueous sodium hydroxide solution (303 mL, 605 mmol) at room temperature.
  • reaction mixture is stirred in a 47 °C heating block overnight with an internal temperature of 45 °C.
  • the reaction mixture is cooled to room temperature and concentrated to remove 2-propanol, then the mixture is diluted with water (50 mL).
  • the mixture is extracted with methyl tert- butyl ether (250 mL), then the aqueous layer is cooled in an ice-water bath and acidified with acetic acid (55.6 mL, 968 mmol).
  • Tetrahydrofuran (798 mL) is added to a flask containing 2-[[(2S,4S)-l-tert- butoxycarbonyl-2-methyl-4-piperidyl]oxy]acetic acid (79.8 g, 224 mmol, 76.6 mass%) and the mixture is stirred in an ice-water bath with an internal temperature of 5 °C.
  • ⁇ , ⁇ -carbonyldiimidazole (43.5 g, 268 mmol) in one portion and the reaction mixture is stirred at room temperature for 2 hours.
  • An additional portion of 1,1'- carbonyldiimidazole (7.25 g, 44.7 mmol) is added and the reaction mixture is stirred at room temperature for 30 minutes.
  • reaction mixture is submerged in an ice-water bath and acetohydrazide (21.5 g, 291 mmol) is added in one portion, then the reaction mixture is stirred at room temperature overnight.
  • the reaction mixture is stirred in an ice-water bath and saturated aqueous sodium bicarbonate solution (500 mL) is added over 2 minutes, maintaining an internal temperature below 15 °C.
  • the mixture is diluted with water (300 mL) and then is concentrated to remove tetrahydrofuran.
  • the aqueous mixture is extracted with 2-methyltetrahydrofuran (4 x 500 mL).
  • N,N-diisopropylethylamine 114 mL, 652 mmol
  • p-toluenesulfonyl chloride 77.7 g, 408 mmol
  • N',N'-dimethylethane-l,2-diamine (21.8 g, 245 mmol) is added dropwise over 10 minutes, maintaining an internal temperature below 15 °C.
  • the reaction mixture is stirred at room temperature for 30 minutes, then is diluted with saturated aqueous citric acid solution (50 mL), ethyl acetate (500 mL) and water (450 mL) at room temperature. The layers are separated and the organic layer is washed with a mixture of saturated aqueous citric acid solution (50 mL) and water (450 mL).
  • N-(4-fluoro-5-formyl-thiazol-2-yl)acetamide (0.04g, 0.122 mmol) is added and stirred for 5 minutes, sodium triacetoxyborohydride (0.055g, 0.25 mmol) is added and reaction mixture is warmed to 40 °C and stirred overnight. The mixture is concentrated under reduced pressure to afford a brown solid.
  • reaction mixture is stirred in a 31 °C heating block for 15 minutes with an internal temperature of 30 °C, then N-(4-fluoro-5-formyl-thiazol-2-yl)acetamide (17.5 g, 93.0 mmol) is added portionwise over 5 minutes.
  • the reaction mixture is stirred in a 31 °C heating block overnight with an internal temperature of 30 °C, then is cooled in an ice-water bath to an internal temperature of 5 °C.
  • To the mixture is added 2M aqueous hydrochloric acid solution (140 mL) over 15 minutes, maintaining an internal temperature below 10 °C.
  • the mixture is stirred at room temperature for 15 minutes, then is diluted with water (50 mL) and ethyl acetate (20 mL) and the layers are separated.
  • the organic layer is extracted with a mixture of 2M aqueous hydrochloric acid solution (35 mL) in water (100 mL).
  • the combined aqueous layers are stirred in an ice-water bath and 50% aqueous sodium hydroxide solution (19.5 mL) is added dropwise over 10 minutes, maintaining an internal temperature below 10 °C.
  • the mixture is diluted with saturated aqueous sodium bicarbonate solution (50 mL), then is extracted with
  • the filtrate is concentrated to give a residue, which is combined with 40% ethyl acetate in heptane (50 mL) and the mixture is stirred in a 50 °C heating block for 30 minutes, then is cooled to room temperature and filtered.
  • the filtered solid is combined with 50% ethyl acetate in heptane (33 mL) and the mixture is stirred in a 50 °C heating block for 1 hour, then is cooled to room temperature and filtered.
  • the filtered solid is dried under vacuum at 40 °C for 1 hour to give a second crop of product (2.50 g).
  • a combination of lots including the first and second crops of product (29.3 g, 76.4 mmol) is combined with ethyl acetate (117 mL) and heptane (117 mL) at room
  • the sample is scanned between 4 and 40° in 2 ⁇ , with a step size of 0.0087° in 2 ⁇ and a scan rate of 0.5 seconds/step, and with 0.6 mm divergence, 5.28mm fixed anti-scatter, and 9.5 mm detector slits.
  • the dry powder is packed on a quartz sample holder and a smooth surface is obtained using a glass slide.
  • peak positions can shift due to a variation in the temperature or humidity at which a sample is analyzed, sample displacement, or the presence or absence of an internal standard.
  • a peak position variability of ⁇ 0.2 in 2 ⁇ will take into account these potential variations without hindering the unequivocal identification of the indicated crystal form.
  • Confirmation of a crystal form may be made based on any unique combination of distinguishing peaks (in units of ° 2 ⁇ ), typically the more prominent peaks.
  • the crystal form diffraction patterns, collected at ambient temperature and relative humidity, were adjusted based on NIST 675 standard peaks at 8.85 and 26.77 degrees 2-theta.
  • crystalline N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,3,4-oxadiazol- 2-yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide is characterized by an XRD pattern using CuKa radiation as having diffraction peaks (2-theta values) as described in Table 1. More specifically, the pattern preferably contains a peak at 13.5° in combination with one or more peaks selected from the group consisting of 5.8°, 13.0°, 14.3°, 17.5°, 20.4°, 21.4°, and 22.2 ° with a tolerance for the diffraction angles of 0.2 degrees.
  • Table 1 X-ray powder diffraction peaks of crystalline N-[4-fluoro-5-[[(2S,4S)-2-methyl- 4-[(5-methyl-l,3,4-oxadiazol-2-yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide.
  • the nucleotide sequence encoding full-length human O-GlcNAc- ⁇ - ⁇ - acetylglucosaminidase ( M_012215) is inserted into pFastBacl (Invitrogen) vector with an N-terminal poly-histidine (HIS) tag.
  • Baculovirus generation is carried out according to the Bac-to-Bac Baculovirus Expression system (Invitrogen) protocol. Sf9 cells are infected at 1.5 x 10 6 cells/mL using 10 mL of PI virus per Liter of culture and incubated at 28 ° C for 48hrs. Cells are spun down, rinsed with PBS and the pellets stored at -80 ° C.
  • the above OGA protein (His-OGA) is purified as follows: 4 L of cells are lysed in 200 mL of buffer containing 50 mM Tris, pH 8.0, 300 mM NaCl, 10% glycerol, 10 mM Imidazol, 1 mM Dithiothreitol (DTT), 0.1% TritonTM X-100, 4 tablets of protease inhibitors (complete EDTA-Free, Roche) for 45 min at 4 ° C. This cell lysate is then spun for 40 min at 16500 rpm at 4 ° C, and supernatant incubated with 6 mL of Ni-NTA resin (nickel-nitrilotriacetic acid) for 2 hours at 4 ° C.
  • Ni-NTA resin nickel-nitrilotriacetic acid
  • Resin is then packed onto column and washed with 50 mM Tris, pH 8.0, 300 mM
  • the protein is eluted with 50 mM Tris, pH 8.0, 150 mM NaCl, 10%) glycerol, 2 mM DTT. Fractions containing His-OGA are pooled and protein concentration measured with BCA (Bradford Colorimetric Assay).
  • the OGA enzyme catalyses the removal of O-GlcNAc from nucleocytoplasmic proteins.
  • Fluorescein di-N-acetyl-P-N-acetyl-D-glucosaminide FD-GlcNAc, Kim, Eun Ju; Kang, Dae Ook; Love, Dona C; Hanover, John A.
  • the assay buffer is prepared to give a final concentration of 50 mM H2NaP03-HNa 2 P0 3 , 0.01% bovine serum albumin and 0.01% TritonTM X-100 in water, at pH 7.
  • the final enzyme concentration is 3 nM (in the 96 well assay format) or 3.24 nM (in the 384 well assay format). Both assay formats yield essentially equivalent results.
  • DMSO dimethyl sulfoxide
  • TRex-293 cells modified for inducible expression of the P301 S-1N4R form of the microtubule associated protein tau are generated and maintained in growth media, consisting of DMEM High Glucose (Sigma# D5796), supplemented with 10 % Tetracyclin-free Fetal Bovine Serum (FBS, Sigma F2442), 20 mM HEPES, 5 ⁇ g/mL Blasticidin (Life Technologies# Al 1139-03) and 200 ⁇ g/mL Zeocin (Life Technologies# R250-01).
  • cells are plated in growth media at 10,000-14,000 cells per well in a Corning Biocoat (356663) 384 well plate coated with poly-D-Lysine, and incubated 20-24 h in a cell incubator at 37°C/5% C0 2 . Experiments are performed without inducing Tau expression.
  • Compounds to be tested are serially diluted 1/3 in pure DMSO using ten point concentration response curves and further diluted in growth media. 20-24 h after plating, cells are treated with test compound in growth media; maximal compound concentration is 15 ⁇ (0.15% DMSO). The maximum inhibition is defined by replicate measurements of 15 uM Thiamet G and the minimum inhibition is defined by replicate measurements of 0.15% DMSO treatment. The cells are returned to the incubator at 37°C/5% C0 2 for 20- 24 hours. Compounds are tested in duplicates within each plate.
  • the media is removed from the assay plate and 25 ⁇ ⁇ of 3.7 % Formaldehyde solution (Sigma # F1635) in DPBS (Sigma #D8537; Dulbecco's phosphate buffered saline) is added to each well and incubated for 30 minutes.
  • DPBS Dulbecco's phosphate buffered saline
  • the cells are then washed once with DPBS and then permeabilized with 0.1% TritonTM X-100 (Sigma# T9284). After 30 minutes, cells are washed twice with DPBS and then blocking solution(l% BSA/DPBS/0.1% TritonTM X-100) is added to each well and incubated for 60 minutes.
  • the blocking solution is removed and a 0.40- 0.33 ⁇ g/mL solution ⁇ ⁇ -GlcNAc Protein antibody (RL2 clone, Thermo, MA1072) in blocking solution is added to the cells and allowed to sit overnight at 2-8 °C. The next day, the cells are washed twice with DPBS and the secondary antibody, Alexa Fluor 488 goat anti-mouse IgG (Life Technologies # Al 1001) at 2 ug/mL in DPBS is added to each well and allowed to sit at room temperature for 90 min.
  • the secondary antibody is removed, cells washed twice with DPBS and a solution of DAPI (Sigma #D9564; 4 ',6- diamidino-2-phenyindole, dilactate) and RNase (Sigma, R6513) in DPBS at a
  • the plates are analyzed on an Acumen eX3 instrument using a 488 and 405 nm excitation lasers and two emission filters FL2 (500-530 nm) and FL1 (420-490 nm).
  • the FL2 filter is the signal corresponding to the O-GlcNAc Protein antibody (RL2 clone) and the FL1 filter is the signal corresponding to the cell nuclei (DAPI).
  • the ratio Total FL2/Total FL1 Total fluorescence of each well without object or population selection) is used for data analysis.
  • the data are normalized to a maximum inhibition as referenced by a 15 ⁇ treatment of Thiamet G and a minimum inhibition as achieved by a 0.15% DMSO treatment.
  • the data are fitted with a non-linear curve fitting application (4- parameters logistic equation) and IC50 values are calculated and reported.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Neurosurgery (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Psychiatry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

La présente invention concerne un composé de formule I : ou un sel pharmaceutiquement acceptable de celui-ci, et l'utilisation de composés de formule I pour le traitement de maladies et de troubles neurodégénératifs, tels que la maladie d'Alzheimer.
PCT/US2018/033368 2017-05-25 2018-05-18 Composés de 5-méthyl-1,3,4-oxadiazol-2-yl WO2018217558A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN201880033585.3A CN110650957B (zh) 2017-05-25 2018-05-18 5-甲基-1,3,4-噁二唑-2-基化合物
EP18729282.6A EP3630755B1 (fr) 2017-05-25 2018-05-18 Composés de 5-méthyl-1,3,4-oxadiazol-2-yl
ES18729282T ES2935349T3 (es) 2017-05-25 2018-05-18 Compuestos de 5-metil-1,3,4-oxadiazol-2-ilo
JP2019564808A JP6810285B2 (ja) 2017-05-25 2018-05-18 5−メチル−1,3,4−オキサジアゾール−2−イル化合物

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762510776P 2017-05-25 2017-05-25
US62/510,776 2017-05-25

Publications (1)

Publication Number Publication Date
WO2018217558A1 true WO2018217558A1 (fr) 2018-11-29

Family

ID=62528882

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/033368 WO2018217558A1 (fr) 2017-05-25 2018-05-18 Composés de 5-méthyl-1,3,4-oxadiazol-2-yl

Country Status (8)

Country Link
US (1) US10377750B2 (fr)
EP (1) EP3630755B1 (fr)
JP (1) JP6810285B2 (fr)
CN (1) CN110650957B (fr)
AR (1) AR111693A1 (fr)
ES (1) ES2935349T3 (fr)
TW (1) TWI669302B (fr)
WO (1) WO2018217558A1 (fr)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10336775B2 (en) 2014-08-28 2019-07-02 Asceneuron Sa Glycosidase inhibitors
US10556902B2 (en) 2016-02-25 2020-02-11 Asceneuron Sa Glycosidase inhibitors
US10696668B2 (en) 2016-02-25 2020-06-30 Asceneuron Sa Acid addition salts of piperazine derivatives
WO2020169804A1 (fr) 2019-02-22 2020-08-27 Asceneuron Sa Inhibiteurs de glycosidases fusionnés
WO2021094312A1 (fr) 2019-11-11 2021-05-20 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga contenant de la pyrrolidine et de la bicyclohétéroaryle
WO2021110656A1 (fr) 2019-12-02 2021-06-10 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
WO2021123291A1 (fr) 2019-12-18 2021-06-24 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
WO2021123294A1 (fr) 2019-12-18 2021-06-24 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
WO2021123297A1 (fr) 2019-12-18 2021-06-24 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
JP2021530552A (ja) * 2018-07-31 2021-11-11 イーライ リリー アンド カンパニー 併用療法
JP2021533105A (ja) * 2018-07-31 2021-12-02 イーライ リリー アンド カンパニー 5−メチル−4−フルオロ−チアゾール−2−イル化合物
US11213525B2 (en) 2017-08-24 2022-01-04 Asceneuron Sa Linear glycosidase inhibitors
US11261183B2 (en) 2016-02-25 2022-03-01 Asceneuron Sa Sulfoximine glycosidase inhibitors
US11612599B2 (en) 2016-02-25 2023-03-28 Asceneuron Sa Glycosidase inhibitors
WO2023070064A1 (fr) * 2021-10-22 2023-04-27 Eli Lilly And Company Polythérapie par un inhibiteur d'o-glcnacase (oga)
US11731972B2 (en) 2018-08-22 2023-08-22 Asceneuron Sa Spiro compounds as glycosidase inhibitors
US11795165B2 (en) 2018-08-22 2023-10-24 Asceneuron Sa Tetrahydro-benzoazepine glycosidase inhibitors
US12016852B2 (en) 2018-08-22 2024-06-25 Asceneuron Sa Pyrrolidine glycosidase inhibitors

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI726329B (zh) 2018-06-22 2021-05-01 美商美國禮來大藥廠 2,3-二氫呋喃并[2,3-b]吡啶化合物
TWI716107B (zh) 2018-09-26 2021-01-11 美商美國禮來大藥廠 6-氟-2-甲基苯并[d]噻唑-5-基化合物

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004094380A1 (fr) 2003-04-18 2004-11-04 Eli Lilly And Company Composes (piperidinyloxy)phenyle, (piperidinyloxy)pyridinyle, (piperidinylsulfanyl)phenyle et (piperidinylsulfanyl)pyridinyle utilises comme agonistes des recepteurs 5-ht1f
US9120781B2 (en) 2010-05-11 2015-09-01 Simon Fraser University Selective glycosidase inhibitors and uses thereof
US20160031871A1 (en) 2013-03-14 2016-02-04 Henry Yu Glycosidase Inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8726763D0 (en) 1987-11-16 1987-12-23 Fujisawa Pharmaceutical Co Thiazole compounds
SE0302116D0 (sv) * 2003-07-21 2003-07-21 Astrazeneca Ab Novel compounds
WO2015046193A1 (fr) * 2013-09-25 2015-04-02 塩野義製薬株式会社 Dérivé amine hétérocyclique aromatique présentant une activité inhibitrice de trpv4
MA53944A (fr) 2014-08-28 2021-08-25 Asceneuron Sa Inhibiteurs de la glycosidase
WO2017106254A1 (fr) 2015-12-18 2017-06-22 Merck Sharp & Dohme Corp. Inhibiteurs de glycosidase et leurs utilisations
AR110747A1 (es) 2017-01-27 2019-05-02 Lilly Co Eli Compuestos de 5-metil-1,2,4-oxadiazol-3-ilo

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004094380A1 (fr) 2003-04-18 2004-11-04 Eli Lilly And Company Composes (piperidinyloxy)phenyle, (piperidinyloxy)pyridinyle, (piperidinylsulfanyl)phenyle et (piperidinylsulfanyl)pyridinyle utilises comme agonistes des recepteurs 5-ht1f
US9120781B2 (en) 2010-05-11 2015-09-01 Simon Fraser University Selective glycosidase inhibitors and uses thereof
US20160031871A1 (en) 2013-03-14 2016-02-04 Henry Yu Glycosidase Inhibitors

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2012, PHARMACEUTICAL PRESS
"The U. S. Pharmacopeia 38 - National Formulary 35", 1 May 2015, article "Characterization of crystalline and partially crystalline solids by X-ray powder diffraction (XRPD) Official"
BASTIN, R.J. ET AL.: "Salt Selection and Optimization Procedures for Pharmaceutical New Chemical Entities", ORGANIC PROCESS RESEARCH AND DEVELOPMENT, vol. 4, 2000, pages 427 - 435
BERGE, S.M. ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
E.L. ELIEL; S.H. WILEN: "Stereochemistry of Organic Compounds", 1994, WILEY-INTERSCIENCE
FAHIMEH MORADI-AFRAPOLI ET AL: "In vitro [alpha]-glucosidase inhibitory activity of phenolic constituents from aerial parts of Polygonum hyrcan", DARU JOURNAL OF PHARMACEUTICAL SCIENCES, BIOMED CENTRAL LTD, LONDON, UK, vol. 20, no. 1, 10 September 2012 (2012-09-10), pages 37, XP021119779, ISSN: 2008-2231, DOI: 10.1186/2008-2231-20-37 *
GOULD, P.L.: "Salt selection for basic drugs", INTERNATIONAL JOURNAL OF PHARMACEUTICS, vol. 33, 1986, pages 201 - 217, XP025813036, DOI: doi:10.1016/0378-5173(86)90055-4
GRAHAM ET AL., NEUROPHARMACOLOGY, vol. 79, 2014, pages 307 - 313
J. JACQUES ET AL.: "Enantiomers, Racemates, and Resolutions", 1981, JOHN WILEY AND SONS, INC.
KIM, EUN JU; KANG, DAE OOK; LOVE, DONA C.; HANOVER; JOHN A, CARBOHYDRATE RESEARCH, vol. 341, no. 8, 2006, pages 971 - 982
N. MASUDA ET AL., BIOORG MED CHEM, vol. 12, 2004, pages 6171 - 6182
NELSON ET AL., JNEUROPATHOL EXP NEUROL., vol. 71, no. 5, 2012, pages 362 - 381
T. W. GREEN; P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY-INTERSCIENCE
WILLIAMS ET AL., BRAIN, vol. 130, 2007, pages 1566 - 76
YAO CHEN ET AL: "Discovery of new acetylcholinesterase and butyrylcholinesterase inhibitors through structure-based virtual screening", RSC ADVANCES, vol. 7, no. 6, 1 January 2017 (2017-01-01), pages 3429 - 3438, XP055488260, DOI: 10.1039/C6RA25887E *
YUZWA ET AL., NAT CHEM BIOL, vol. 4, no. 8, 2008, pages 483 - 490
YUZWA ET AL., NAT CHEM BIOL, vol. 8, 2012, pages 393 - 399

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11046712B2 (en) 2014-08-28 2021-06-29 Asceneuron Sa Glycosidase inhibitors
US10336775B2 (en) 2014-08-28 2019-07-02 Asceneuron Sa Glycosidase inhibitors
US10556902B2 (en) 2016-02-25 2020-02-11 Asceneuron Sa Glycosidase inhibitors
US10696668B2 (en) 2016-02-25 2020-06-30 Asceneuron Sa Acid addition salts of piperazine derivatives
US11612599B2 (en) 2016-02-25 2023-03-28 Asceneuron Sa Glycosidase inhibitors
US10995090B2 (en) 2016-02-25 2021-05-04 Asceneuron Sa Substituted dihydrobenzofuran glycosidase inhibitors
US11591327B2 (en) 2016-02-25 2023-02-28 Asceneuron Sa Acid addition salts of piperazine derivatives
US11261183B2 (en) 2016-02-25 2022-03-01 Asceneuron Sa Sulfoximine glycosidase inhibitors
US11213525B2 (en) 2017-08-24 2022-01-04 Asceneuron Sa Linear glycosidase inhibitors
JP2021533105A (ja) * 2018-07-31 2021-12-02 イーライ リリー アンド カンパニー 5−メチル−4−フルオロ−チアゾール−2−イル化合物
JP2021530552A (ja) * 2018-07-31 2021-11-11 イーライ リリー アンド カンパニー 併用療法
JP7016446B2 (ja) 2018-07-31 2022-02-04 イーライ リリー アンド カンパニー 5-メチル-4-フルオロ-チアゾール-2-イル化合物
JP2022191382A (ja) * 2018-07-31 2022-12-27 イーライ リリー アンド カンパニー 併用療法
JP7361179B2 (ja) 2018-07-31 2023-10-13 イーライ リリー アンド カンパニー 併用療法
US11731972B2 (en) 2018-08-22 2023-08-22 Asceneuron Sa Spiro compounds as glycosidase inhibitors
US12016852B2 (en) 2018-08-22 2024-06-25 Asceneuron Sa Pyrrolidine glycosidase inhibitors
US11795165B2 (en) 2018-08-22 2023-10-24 Asceneuron Sa Tetrahydro-benzoazepine glycosidase inhibitors
WO2020169804A1 (fr) 2019-02-22 2020-08-27 Asceneuron Sa Inhibiteurs de glycosidases fusionnés
WO2021094312A1 (fr) 2019-11-11 2021-05-20 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga contenant de la pyrrolidine et de la bicyclohétéroaryle
WO2021110656A1 (fr) 2019-12-02 2021-06-10 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
WO2021123291A1 (fr) 2019-12-18 2021-06-24 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
WO2021123297A1 (fr) 2019-12-18 2021-06-24 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
WO2021123294A1 (fr) 2019-12-18 2021-06-24 Janssen Pharmaceutica Nv Composés inhibiteurs d'oga
WO2023070064A1 (fr) * 2021-10-22 2023-04-27 Eli Lilly And Company Polythérapie par un inhibiteur d'o-glcnacase (oga)

Also Published As

Publication number Publication date
AR111693A1 (es) 2019-08-07
ES2935349T3 (es) 2023-03-06
US10377750B2 (en) 2019-08-13
TW201904963A (zh) 2019-02-01
EP3630755A1 (fr) 2020-04-08
JP6810285B2 (ja) 2021-01-06
TWI669302B (zh) 2019-08-21
CN110650957A (zh) 2020-01-03
CN110650957B (zh) 2022-12-09
US20180339984A1 (en) 2018-11-29
EP3630755B1 (fr) 2022-12-14
JP2020520962A (ja) 2020-07-16

Similar Documents

Publication Publication Date Title
EP3630755B1 (fr) Composés de 5-méthyl-1,3,4-oxadiazol-2-yl
AU2018213029B2 (en) N-(4-fluoro-5-(((2s,4s)-2-methyl-4-((5-methyl-1,2,4-oxadiazol-3-yl)methoxy)-1-piperidyl)methyl)thiazol-2-yl)acetamide as oga inhibitor
KR20080047581A (ko) 인자 xa 억제제로서의 신규 사이클로알케인카복스아마이드
WO2012037351A1 (fr) Composés
FR2758329A1 (fr) Derives d'imidazole-4-butane boronique, leur preparation et leur utilisation en therapeutique
NZ754849B2 (en) N-[4-fluoro-5-[[(2s,4s)-2-methyl-4-[(5-methyl-1,2,4-oxadiazol-3-yl)methoxy]-1-piperidyl]methyl]thiazol-2-yl]acetamide as oga inhibitor
CA3108158C (fr) Composes 5-methyl-4-fluoro-thiazol-2-yl
US20200207782A1 (en) Chemically activated water-soluble prodrug
RU2762189C2 (ru) Соли и кристаллические формы диазабензофлуорантреновых соединений

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18729282

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019564808

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2018729282

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2018729282

Country of ref document: EP

Effective date: 20200102